Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
PLoS Pathog ; 11(4): e1004818, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25927232

RESUMEN

Uropathogenic E. coli (UPEC) is the primary cause of urinary tract infections (UTI) affecting approximately 150 million people worldwide. Here, we revealed the importance of transcriptional regulator hypoxia-inducible factor-1 α subunit (HIF-1α) in innate defense against UPEC-mediated UTI. The effects of AKB-4924, a HIF-1α stabilizing agent, were studied using human uroepithelial cells (5637) and a murine UTI model. UPEC adherence and invasion were significantly reduced in 5637 cells when HIF-1α protein was allowed to accumulate. Uroepithelial cells treated with AKB-4924 also experienced reduced cell death and exfoliation upon UPEC challenge. In vivo, fewer UPEC were recovered from the urine, bladders and kidneys of mice treated transurethrally with AKB-4924, whereas increased bacteria were recovered from bladders of mice with a HIF-1α deletion. Bladders and kidneys of AKB-4924 treated mice developed less inflammation as evidenced by decreased pro-inflammatory cytokine release and neutrophil activity. AKB-4924 impairs infection in uroepithelial cells and bladders, and could be correlated with enhanced production of nitric oxide and antimicrobial peptides cathelicidin and ß-defensin-2. We conclude that HIF-1α transcriptional regulation plays a key role in defense of the urinary tract against UPEC infection, and that pharmacological HIF-1α boosting could be explored further as an adjunctive therapy strategy for serious or recurrent UTI.


Asunto(s)
Infecciones por Escherichia coli/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunidad Innata , Infecciones Urinarias/metabolismo , Escherichia coli Uropatógena/inmunología , Urotelio/metabolismo , Administración Intravesical , Animales , Antibacterianos/administración & dosificación , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Péptidos Catiónicos Antimicrobianos/agonistas , Péptidos Catiónicos Antimicrobianos/metabolismo , Adhesión Bacteriana/efectos de los fármacos , Línea Celular , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/prevención & control , Femenino , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/agonistas , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Inmunidad Innata/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/agonistas , Óxido Nítrico/metabolismo , Piperazinas/administración & dosificación , Piperazinas/farmacología , Piperazinas/uso terapéutico , Estabilidad Proteica/efectos de los fármacos , Piridonas/administración & dosificación , Piridonas/farmacología , Piridonas/uso terapéutico , ARN Mensajero/metabolismo , Infecciones Urinarias/inmunología , Infecciones Urinarias/microbiología , Infecciones Urinarias/prevención & control , Escherichia coli Uropatógena/efectos de los fármacos , Urotelio/efectos de los fármacos , Urotelio/inmunología , Urotelio/microbiología
2.
PLoS Pathog ; 10(1): e1003846, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24391502

RESUMEN

Group B Streptococcus (GBS) is a common agent of bacterial sepsis and meningitis in newborns. The GBS surface capsule contains sialic acids (Sia) that engage Sia-binding immunoglobulin-like lectins (Siglecs) on leukocytes. Here we use mice lacking Siglec-E, an inhibitory Siglec of myelomonocytic cells, to study the significance of GBS Siglec engagement during in vivo infection. We found GBS bound to Siglec-E in a Sia-specific fashion to blunt NF-κB and MAPK activation. As a consequence, Siglec-E-deficient macrophages had enhanced pro-inflammatory cytokine secretion, phagocytosis and bactericidal activity against the pathogen. Following pulmonary or low-dose intravenous GBS challenge, Siglec-E KO mice produced more pro-inflammatory cytokines and exhibited reduced GBS invasion of the central nervous system. In contrast, upon high dose lethal challenges, cytokine storm in Siglec-E KO mice was associated with accelerated mortality. We conclude that GBS Sia mimicry influences host innate immune and inflammatory responses in vivo through engagement of an inhibitory Siglec, with the ultimate outcome of the host response varying depending upon the site, stage and magnitude of infection.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos B/inmunología , Imitación Molecular/inmunología , Ácido N-Acetilneuramínico/inmunología , Neumonía Bacteriana/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus agalactiae/inmunología , Animales , Antígenos CD/genética , Antígenos de Diferenciación de Linfocitos B/genética , Citocinas/genética , Citocinas/inmunología , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Noqueados , Ácido N-Acetilneuramínico/genética , Neumonía Bacteriana/genética , Neumonía Bacteriana/patología , Infecciones Estreptocócicas/genética , Infecciones Estreptocócicas/patología
3.
J Biol Chem ; 288(9): 6417-26, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23322784

RESUMEN

Staphylococcus aureus infections present an enormous global health concern complicated by an alarming increase in antibiotic resistance. S. aureus is among the few bacterial species that express nitric-oxide synthase (bNOS) and thus can catalyze NO production from L-arginine. Here we generate an isogenic bNOS-deficient mutant in the epidemic community-acquired methicillin-resistant S. aureus (MRSA) USA300 clone to study its contribution to virulence and antibiotic susceptibility. Loss of bNOS increased MRSA susceptibility to reactive oxygen species and host cathelicidin antimicrobial peptides, which correlated with increased MRSA killing by human neutrophils and within neutrophil extracellular traps. bNOS also promoted resistance to the pharmaceutical antibiotics that act on the cell envelope such as vancomycin and daptomycin. Surprisingly, bNOS-deficient strains gained resistance to aminoglycosides, suggesting that the role of bNOS in antibiotic susceptibility is more complex than previously observed in Bacillus species. Finally, the MRSA bNOS mutant showed reduced virulence with decreased survival and smaller abscess generation in a mouse subcutaneous infection model. Together, these data indicate that bNOS contributes to MRSA innate immune and antibiotic resistance phenotypes. Future development of specific bNOS inhibitors could be an attractive option to simultaneously reduce MRSA pathology and enhance its susceptibility to commonly used antibiotics.


Asunto(s)
Proteínas Bacterianas/metabolismo , Resistencia a la Meticilina , Staphylococcus aureus Resistente a Meticilina/enzimología , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Óxido Nítrico Sintasa/metabolismo , Infecciones Cutáneas Estafilocócicas/enzimología , Absceso/genética , Absceso/microbiología , Absceso/patología , Animales , Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Proteínas Bacterianas/genética , Daptomicina/farmacología , Humanos , Staphylococcus aureus Resistente a Meticilina/genética , Ratones , Mutación , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/genética , Especies Reactivas de Oxígeno/metabolismo , Infecciones Cutáneas Estafilocócicas/genética , Infecciones Cutáneas Estafilocócicas/microbiología , Infecciones Cutáneas Estafilocócicas/patología , Vancomicina/farmacología , Catelicidinas
4.
Sci Rep ; 13(1): 13524, 2023 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-37598239

RESUMEN

The NLRP3 inflammasome is an intracellular, multiprotein complex that promotes the auto-catalytic activation of caspase-1 and the subsequent maturation and secretion of the pro-inflammatory cytokines, IL-1ß and IL-18. Persistent activation of the NLRP3 inflammasome has been implicated in the pathophysiology of a number of inflammatory and autoimmune diseases, including neuroinflammation, cardiovascular disease, non-alcoholic steatohepatitis, lupus nephritis and severe asthma. Here we describe the preclinical profile of JT002, a novel small molecule inhibitor of the NLRP3 inflammasome. JT002 potently reduced NLRP3-dependent proinflammatory cytokine production across a number of cellular assays and prevented pyroptosis, an inflammatory form of cell death triggered by active caspase-1. JT002 demonstrated in vivo target engagement at therapeutically relevant concentrations when orally dosed in mice and prevented body weight loss and improved inflammatory and fibrotic endpoints in a model of Muckle-Wells syndrome (MWS). In two distinct models of neutrophilic airway inflammation, JT002 treatment significantly reduced airway hyperresponsiveness and airway neutrophilia. These results provide a rationale for the therapeutic targeting of the NLRP3 inflammasome in severe asthma and point to the use of JT002 in a variety of inflammatory disorders.


Asunto(s)
Enfermedades Cardiovasculares , Nefritis Lúpica , Animales , Ratones , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Caspasa 1
5.
J Bacteriol ; 194(2): 509-17, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22081398

RESUMEN

Miniature inverted terminal repeat elements (MITEs) are nonautonomous mobile elements that have a significant impact on bacterial evolution. Here we characterize E622, a 611-bp virulence-associated MITE from Pseudomonas syringae, which contains no coding region but has almost perfect 168-bp inverted repeats. Using an antibiotic coupling assay, we show that E622 is transposable and can mobilize an antibiotic resistance gene contained between its borders. Its predicted parent element, designated TnE622, has a typical transposon structure with a three-gene operon, consisting of resolvase, integrase, and exeA-like genes, which is bounded by the same terminal inverted repeats as E622. A broader genome level survey of the E622/TnE622 inverted repeats identified homologs in Pseudomonas, Salmonella, Shewanella, Erwinia, Pantoea, and the cyanobacteria Nostoc and Cyanothece, many of which appear to encompass known virulence genes, including genes encoding toxins, enzymes, and type III secreted effectors. Its association with niche-specific genetic determinants, along with its persistence and evolutionary diversification, indicates that this mobile element family has played a prominent role in the evolution of many agriculturally and clinically relevant pathogenic bacteria.


Asunto(s)
Secuencias Repetitivas Esparcidas/genética , Pseudomonas syringae/genética , Pseudomonas syringae/patogenicidad , Secuencias Repetidas Terminales/genética , Cianobacterias/genética , Cianobacterias/metabolismo , ADN Bacteriano/genética , Genoma Bacteriano , Datos de Secuencia Molecular , Hibridación de Ácido Nucleico , Filogenia , Virulencia
6.
Infect Immun ; 79(6): 2345-55, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21402762

RESUMEN

Staphylococcus aureus is a frequent cause of bloodstream, respiratory tract, and skin and soft tissue infections. In the bloodstream, the iron-binding glycoprotein transferrin circulates to provide iron to cells throughout the body, but its iron-binding properties make it an important component of innate immunity. It is well established that siderophores, with their high affinity for iron, in many instances can remove iron from transferrin as a means to promote proliferation of bacterial pathogens. It is also established that catecholamine hormones can interfere with the iron-binding properties of transferrin, thus allowing infectious bacteria access to this iron pool. The present study demonstrates that S. aureus can use either of two carboxylate-type siderophores, staphyloferrin A and staphyloferrin B, via the transporters Hts and Sir, respectively, to access the transferrin iron pool. Growth of staphyloferrin-producing S. aureus in serum or in the presence of holotransferrin was not enhanced in the presence of catecholamines. However, catecholamines significantly enhanced the growth of staphyloferrin-deficient S. aureus in human serum or in the presence of human holotransferrin. It was further demonstrated that the Sst transporter was essential for this activity as well as for the utilization of bacterial catechol siderophores. The substrate binding protein SstD was shown to interact with ferrated catecholamines and catechol siderophores, with low to submicromolar affinities. Experiments involving mice challenged intravenously with wild-type S. aureus and isogenic mutants demonstrated that the combination of Hts, Sir, and Sst transport systems was required for full virulence of S. aureus.


Asunto(s)
Citratos/fisiología , Hierro/fisiología , Ornitina/análogos & derivados , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/patogenicidad , Transferrina/metabolismo , Animales , Western Blotting , Citratos/metabolismo , Epinefrina/metabolismo , Epinefrina/fisiología , Femenino , Genes Bacterianos/genética , Genes Bacterianos/fisiología , Humanos , Hierro/metabolismo , Ratones , Ratones Endogámicos BALB C , Norepinefrina/metabolismo , Norepinefrina/fisiología , Ornitina/metabolismo , Ornitina/fisiología , Sideróforos/metabolismo , Sideróforos/fisiología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/metabolismo , Staphylococcus aureus/fisiología
7.
BMC Microbiol ; 11: 199, 2011 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-21906287

RESUMEN

BACKGROUND: Staphylococcus aureus synthesizes two siderophores, staphyloferrin A and staphyloferrin B, that promote iron-restricted growth. Previous work on the biosynthesis of staphyloferrin B has focused on the role of the synthetase enzymes, encoded from within the sbnA-I operon, which build the siderophore from the precursor molecules citrate, alpha-ketoglutarate and L-2,3-diaminopropionic acid. However, no information yet exists on several other enzymes, expressed from the biosynthetic cluster, that are thought to be involved in the synthesis of the precursors (or synthetase substrates) themselves. RESULTS: Using mutants carrying insertions in sbnA and sbnB, we show that these two genes are essential for the synthesis of staphyloferrin B, and that supplementation of the growth medium with L-2,3-diaminopropionic acid can bypass the block in staphyloferrin B synthesis displayed by the mutants. Several mechanisms are proposed for how the enzymes SbnA, with similarity to cysteine synthase enzymes, and SbnB, with similarity to amino acid dehydrogenases and ornithine cyclodeaminases, function together in the synthesis of this unusual nonproteinogenic amino acid L-2,3-diaminopropionic acid. CONCLUSIONS: Mutation of either sbnA or sbnB result in abrogation of synthesis of staphyloferrin B, a siderophore that contributes to iron-restricted growth of S. aureus. The loss of staphyloferrin B synthesis is due to an inability to synthesize the unusual amino acid L-2,3-diaminopropionic acid which is an important, iron-liganding component of the siderophore structure. It is proposed that SbnA and SbnB function together as an L-Dap synthase in the S. aureus cell.


Asunto(s)
Amoníaco-Liasas/genética , Proteínas Bacterianas/genética , Vías Biosintéticas , Citratos/biosíntesis , Cisteína Sintasa/genética , Mutación , Staphylococcus aureus/enzimología , beta-Alanina/análogos & derivados , Amoníaco-Liasas/metabolismo , Proteínas Bacterianas/metabolismo , Cisteína Sintasa/metabolismo , Regulación hacia Abajo , Operón , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , beta-Alanina/biosíntesis
8.
Infect Immun ; 78(4): 1618-28, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20100857

RESUMEN

The tremendous success of Staphylococcus aureus as a pathogen is due to the controlled expression of a diverse array of virulence factors. The effects of host environments on the expression of virulence factors and the mechanisms by which S. aureus adapts to colonize distinct host tissues are largely unknown. Vertebrates have evolved to sequester nutrient iron from invading bacteria, and iron availability is a signal that alerts pathogenic microorganisms when they enter the hostile host environment. Consistent with this, we report here that S. aureus senses alterations in the iron status via the ferric uptake regulator (Fur) and alters the abundance of a large number of virulence factors. These Fur-mediated changes protect S. aureus against killing by neutrophils, and Fur is required for full staphylococcal virulence in a murine model of infection. A potential mechanistic explanation for the impact of Fur on virulence is provided by the observation that Fur coordinates the reciprocal expression of cytolysins and a subset of immunomodulatory proteins. More specifically, S. aureus lacking fur exhibits decreased expression of immunomodulatory proteins and increased expression of cytolysins. These findings reveal that Fur is involved in initiating a regulatory program that organizes the expression of virulence factors during the pathogenesis of S. aureus pneumonia.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Regulación Bacteriana de la Expresión Génica , Neumonía Estafilocócica/microbiología , Proteínas Represoras/fisiología , Staphylococcus aureus/patogenicidad , Factores de Virulencia/biosíntesis , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/fisiología , Cromatografía Liquida , Modelos Animales de Enfermedad , Electroforesis en Gel Bidimensional , Femenino , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Proteoma/análisis , Proteínas Represoras/genética , Staphylococcus aureus/fisiología
9.
Mol Microbiol ; 74(3): 594-608, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19775248

RESUMEN

Siderophores are iron-scavenging molecules produced by many microbes. In general, they are synthesized using either non-ribosomal peptide synthetase (NRPS) or NRPS-independent siderophore (NIS) pathways. Staphylococcus aureus produces siderophores, of which the structures of staphyloferrin A and staphyloferrin B are known. Recently, the NIS biosynthetic pathway for staphyloferrin A was characterized. Here we show that, in S. aureus, the previously identified sbn (siderophore biosynthesis) locus encodes enzymes required for the synthesis of staphyloferrin B, an alpha-hydroxycarboxylate siderophore comprised of l-2,3-diaminopropionic acid, citric acid, 1,2-diaminoethane and alpha-ketoglutaric acid. Staphyloferrin B NIS biosynthesis was recapitulated in vitro, using purified recombinant Sbn enzymes and the component substrates. In vitro synthesized staphyloferrin B readily promoted the growth of iron-starved S. aureus, via the ABC transporter SirABC. The SbnCEF synthetases and a decarboxylase, SbnH, were necessary and sufficient to produce staphyloferrin B in reactions containing component substrates l-2,3-diaminopropionic acid, citric acid and alpha-ketoglutaric acid. Since 1,2-diaminoethane was not required, this component of the siderophore arises from the SbnH-dependent decarboxylation of a 2,3-diaminoproprionic acid-containing intermediate. Liquid chromatography-electrospray ionization-mass spectrometry (LC-ESI-MS) analyses of a series of enzyme reactions identified mass ions corresponding to biosynthetic intermediates, allowing for the first proposed biosynthetic pathway for staphyloferrin B.


Asunto(s)
Citratos/biosíntesis , Ornitina/análogos & derivados , Staphylococcus aureus/metabolismo , Transportadoras de Casetes de Unión a ATP/biosíntesis , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Citratos/química , Citratos/metabolismo , Ácido Cítrico/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Etilenodiaminas/metabolismo , Compuestos Férricos/metabolismo , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos , Hierro/metabolismo , Quelantes del Hierro/metabolismo , Ácidos Cetoglutáricos/metabolismo , Familia de Multigenes , Ornitina/biosíntesis , Péptido Sintasas/biosíntesis , Péptido Sintasas/genética , Péptido Sintasas/metabolismo , Sideróforos/biosíntesis , Sideróforos/genética , Sideróforos/metabolismo , Staphylococcus aureus/genética , Estereoisomerismo , beta-Alanina/análogos & derivados , beta-Alanina/metabolismo
10.
Mol Microbiol ; 72(4): 947-63, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19400778

RESUMEN

Iron is critical for virtually all forms of life. The production of high-affinity iron chelators, siderophores, and the subsequent uptake of iron-siderophore complexes are a common strategy employed by microorganisms to acquire iron. Staphylococcus aureus produces siderophores but genetic information underlying their synthesis and transport is limited. Previous work implicated the sbn operon in siderophore synthesis and the sirABC operon in uptake. Here we characterize a second siderophore biosynthetic locus in S. aureus; the locus consists of four genes (in strain Newman these open reading frames are designated NWMN_2079-2082) which, together, are responsible for the synthesis and export of staphyloferrin A, a polycarboxylate siderophore. While deletion of the NWMN_2079-2082 locus did not affect iron-restricted growth of S. aureus, strains bearing combined sbn and NWMN_2079-2082 locus deletions produced no detectable siderophore and demonstrated severely attenuated iron-restricted growth. Adjacent to NWMN_2079-2082 resides the htsABC operon, encoding an ABC transporter previously implicated in haem acquisition. We provide evidence here that HtsABC, along with the FhuC ATPase, is required for the uptake of staphyloferrin A. The crystal structure of apo-HtsA was determined and identified a large positively charged region in the substrate-binding pocket, in agreement with a role in binding of anionic staphyloferrin A.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas Bacterianas/metabolismo , Citratos/biosíntesis , Ornitina/análogos & derivados , Sideróforos/metabolismo , Staphylococcus aureus/genética , Transportadoras de Casetes de Unión a ATP/genética , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos , Hierro/metabolismo , Sistemas de Lectura Abierta , Operón , Ornitina/biosíntesis , Estructura Terciaria de Proteína , Eliminación de Secuencia , Sideróforos/genética , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/metabolismo
11.
Am J Nephrol ; 28(6): 879-89, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18566542

RESUMEN

BACKGROUND: Early upregulation of receptor-interacting protein-2 (RIP2) expression during peritoneal dialysis (PD)-associated peritonitis correlates with a favorable clinical outcome, while failure to upregulate RIP2 correlates with a protracted course. We noticed that patients who do not upregulate RIP2 during PD-associated peritonitis have more peritoneal macrophages during the early phase of infection. METHODS: To study the mechanism behind this observation, we examined the role of RIP2 in the immune response to bacterial challenge in a mouse model of acute peritonitis. We injected RIP2(+/+) and RIP2(-/-) mice intraperitoneally with a Staphylococcus epidermidis cell free-preparation, and peritoneal cells were isolated 3, 6 and 24 h after challenge. RESULTS: Surprisingly, RIP2(-/-) mice had a comparable influx of inflammatory leukocytes, but had a significantly higher number of peritoneal macrophages at 3 h, indicating delayed emigration of these cells. No significant differences were seen at later times suggesting that migration was delayed but not inhibited. In addition, RIP2(-/-) macrophages were more permissive to intracellular infection by Staphylococcus aureus, indicating that, in the absence of RIP2, resident peritoneal macrophages could become reservoirs of bacteria. CONCLUSION: These findings provide a mechanism for the observation that upregulation of RIP2 expression is required for rapid resolution of peritonitis, by decreasing intracellular infection and by regulating the migration of antigen-presenting cells in the early stages of an inflammatory response.


Asunto(s)
Macrófagos/citología , Diálisis Peritoneal/efectos adversos , Peritonitis/complicaciones , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/deficiencia , Animales , Movimiento Celular , Sistema Libre de Células , Humanos , Infecciones/metabolismo , Inflamación , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Modelos Biológicos , Staphylococcus epidermidis/metabolismo , Factores de Tiempo
13.
PLoS Negl Trop Dis ; 11(2): e0005373, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28158186

RESUMEN

Cryptosporidiosis has emerged as a leading cause of non-viral diarrhea in children under five years of age in the developing world, yet the current standard of care to treat Cryptosporidium infections, nitazoxanide, demonstrates limited and immune-dependent efficacy. Given the lack of treatments with universal efficacy, drug discovery efforts against cryptosporidiosis are necessary to find therapeutics more efficacious than the standard of care. To date, cryptosporidiosis drug discovery efforts have been limited to a few targeted mechanisms in the parasite and whole cell phenotypic screens against small, focused collections of compounds. Using a previous screen as a basis, we initiated the largest known drug discovery effort to identify novel anticryptosporidial agents. A high-content imaging assay for inhibitors of Cryptosporidium parvum proliferation within a human intestinal epithelial cell line was miniaturized and automated to enable high-throughput phenotypic screening against a large, diverse library of small molecules. A screen of 78,942 compounds identified 12 anticryptosporidial hits with sub-micromolar activity, including clofazimine, an FDA-approved drug for the treatment of leprosy, which demonstrated potent and selective in vitro activity (EC50 = 15 nM) against C. parvum. Clofazimine also displayed activity against C. hominis-the other most clinically-relevant species of Cryptosporidium. Importantly, clofazimine is known to accumulate within epithelial cells of the small intestine, the primary site of Cryptosporidium infection. In a mouse model of acute cryptosporidiosis, a once daily dosage regimen for three consecutive days or a single high dose resulted in reduction of oocyst shedding below the limit detectable by flow cytometry. Recently, a target product profile (TPP) for an anticryptosporidial compound was proposed by Huston et al. and highlights the need for a short dosing regimen (< 7 days) and formulations for children < 2 years. Clofazimine has a long history of use and has demonstrated a good safety profile for a disease that requires chronic dosing for a period of time ranging 3-36 months. These results, taken with clofazimine's status as an FDA-approved drug with over four decades of use for the treatment of leprosy, support the continued investigation of clofazimine both as a new chemical tool for understanding cryptosporidium biology and a potential new treatment of cryptosporidiosis.


Asunto(s)
Antiprotozoarios/farmacología , Clofazimina/farmacología , Criptosporidiosis/tratamiento farmacológico , Cryptosporidium parvum/efectos de los fármacos , Reposicionamiento de Medicamentos , Animales , Automatización de Laboratorios , Línea Celular , Criptosporidiosis/parasitología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Células Epiteliales/parasitología , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Resultado del Tratamiento
14.
mBio ; 6(2): e00133, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25759502

RESUMEN

UNLABELLED: The M1T1 clone of group A Streptococcus (GAS) is associated with severe invasive infections, including necrotizing fasciitis and septicemia. During invasive M1T1 GAS disease, mutations in the covRS regulatory system led to upregulation of an ADP-ribosyltransferase, SpyA. Surprisingly, a GAS ΔspyA mutant was resistant to killing by macrophages and caused higher mortality with impaired bacterial clearance in a mouse intravenous challenge model. GAS expression of SpyA triggered macrophage cell death in association with caspase-1-dependent interleukin 1ß (IL-1ß) production, and differences between wild-type (WT) and ΔspyA GAS macrophage survival levels were lost in cells lacking caspase-1, NOD-like receptor protein 3 (NLRP3), apoptosis-associated speck-like protein (ASC), or pro-IL-1ß. Similar in vitro findings were identified in macrophage studies performed with pseudomonal exotoxin A, another ADP-ribosylating toxin. Thus, SpyA triggers caspase-1-dependent inflammatory cell death in macrophages, revealing a toxin-triggered IL-1ß-dependent innate immune response pathway critical in defense against invasive bacterial infection. IMPORTANCE: Group A Streptococcus (GAS) is a leading human pathogen capable of producing invasive infections even in healthy individuals. GAS bacteria produce a toxin called SpyA that modifies host proteins through a process called ADP ribosylation. We describe how macrophages, frontline defenders of the host innate immune system, respond to SpyA by undergoing a specialized form of cell death in which they are activated to release the proinflammatory cytokine molecule interleukin 1ß (IL-1ß). Release of IL-1ß activates host immune cell clearance of GAS, as we demonstrated in tissue culture models of macrophage bacterial killing and in vivo mouse infectious-challenge experiments. Similar macrophage responses to a related toxin of Pseudomonas bacteria were also shown. Thus, macrophages recognize certain bacterial toxins to activate a protective immune response in the host.


Asunto(s)
ADP Ribosa Transferasas/inmunología , Interleucina-1beta/metabolismo , Macrófagos/inmunología , Macrófagos/microbiología , Streptococcus pyogenes/enzimología , Streptococcus pyogenes/inmunología , ADP Ribosa Transferasas/genética , Animales , Supervivencia Celular , Modelos Animales de Enfermedad , Eliminación de Gen , Ratones , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/genética , Virulencia
15.
Chem Biol ; 22(6): 785-92, 2015 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-26091171

RESUMEN

Bacterial infections associated with methicillin-resistant Staphylococcus aureus (MRSA) are a major economic burden to hospitals, and confer high rates of morbidity and mortality among those infected. Exploitation of novel therapeutic targets is thus necessary to combat this dangerous pathogen. Here, we report on the identification and characterization, including crystal structures, of two nitric oxide synthase (NOS) inhibitors that function as antimicrobials against MRSA. These data provide the first evidence that bacterial NOS (bNOS) inhibitors can work synergistically with oxidative stress to enhance MRSA killing. Crystal structures show that each inhibitor contacts an active site Ile residue in bNOS that is Val in the mammalian NOS isoforms. Mutagenesis studies show that the additional nonpolar contacts provided by the Ile in bNOS contribute to tighter binding toward the bacterial enzyme.


Asunto(s)
Proteínas Bacterianas/metabolismo , Staphylococcus aureus Resistente a Meticilina/enzimología , Óxido Nítrico Sintasa/metabolismo , Animales , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Sitios de Unión , Línea Celular , Supervivencia Celular/efectos de los fármacos , Bases de Datos de Proteínas , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/toxicidad , Cinética , Ratones , Simulación del Acoplamiento Molecular , Mutagénesis , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/genética , Unión Proteica , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/metabolismo
16.
J Innate Immun ; 6(4): 485-98, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24481289

RESUMEN

Signaling through Toll-like receptors (TLRs), crucial molecules in the induction of host defense responses, requires adaptor proteins that contain a Toll/interleukin-1 receptor (TIR) domain. The pathogen Staphylococcus aureus produces several innate immune-evasion molecules that interfere with the host's innate immune response. A database search analysis suggested the presence of a gene encoding a homologue of the human TIR domain in S. aureus MSSA476 which was named staphylococcal TIR domain protein (TirS). Ectopic expression of TirS in human embryonic kidney, macrophage and keratinocyte cell lines interfered with signaling through TLR2, including MyD88 and TIRAP, NF-κB and/or mitogen-activated protein kinase pathways. Moreover, the presence of TirS reduced the levels of cytokines MCP-1 and G-CSF secreted in response to S. aureus. The effects on NF-κB pathway were confirmed using S. aureus MSSA476 wild type, an isogenic mutant MSSA476ΔtirS, and complemented MSSA476ΔtirS +pTirS in a Transwell system where bacteria and host cells were physically separated. Finally, in a systematic mouse infection model, TirS promoted bacterial accumulation in several organs 4 days postinfection. The results of this study reveal a new S. aureus virulence factor that can interfere with PAMP-induced innate immune signaling in vitro and bacterial survival in vivo.


Asunto(s)
Proteínas Bacterianas/metabolismo , FN-kappa B/metabolismo , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Receptor Toll-Like 2/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Secuencia de Aminoácidos , Animales , Carga Bacteriana/genética , Proteínas Bacterianas/genética , Línea Celular , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Femenino , Factor Estimulante de Colonias de Granulocitos/metabolismo , Humanos , Evasión Inmune , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Mutación/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Receptores de Interleucina-1/metabolismo , Homología de Secuencia de Aminoácido , Transducción de Señal/genética , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/patogenicidad , Factores de Virulencia/genética
17.
J Inorg Biochem ; 104(3): 282-8, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19850350

RESUMEN

Iron is frequently a growth-limiting nutrient due to its propensity to interact with oxygen to form insoluble precipitates and, therefore, biological systems have evolved specialized uptake mechanisms to obtain this essential nutrient. Many pathogenic bacteria are capable of obtaining stringently sequestered iron from animal hosts by one or both of the following mechanisms: extraction of heme from host erythrocyte and serum hemoproteins, or through the use of high affinity, iron-scavenging molecules termed siderophores. This review summarizes our current knowledge of siderophore-mediated iron acquisition systems in the genus Staphylococcus.


Asunto(s)
Proteínas Bacterianas/metabolismo , Hierro/metabolismo , Sideróforos/metabolismo , Staphylococcus/metabolismo , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Citratos/química , Citratos/metabolismo , Humanos , Modelos Moleculares , Estructura Molecular , Ornitina/análogos & derivados , Ornitina/química , Ornitina/genética , Ornitina/metabolismo , Conformación Proteica , Sideróforos/química , Sideróforos/genética , Staphylococcus/genética
18.
Science ; 329(5989): 294-6, 2010 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-20522739

RESUMEN

Staphylococcus aureus is a major human pathogen that is resistant to numerous antibiotics in clinical use. We found two nonribosomal peptide secondary metabolites--the aureusimines, made by S. aureus--that are not antibiotics, but function as regulators of virulence factor expression and are necessary for productive infections. In vivo mouse models of bacteremia showed that strains of S. aureus unable to produce aureusimines were attenuated and/or cleared from major organs, including the spleen, liver, and heart. Targeting aureusimine synthesis may offer novel leads for anti-infective drugs.


Asunto(s)
Pirazinas/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidad , Factores de Virulencia/metabolismo , Secuencia de Aminoácidos , Animales , Bacteriemia/microbiología , Dipéptidos/química , Dipéptidos/aislamiento & purificación , Corazón/microbiología , Hemólisis , Hígado/microbiología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Biosíntesis de Péptidos Independientes de Ácidos Nucleicos , Péptido Sintasas/química , Péptido Sintasas/genética , Péptido Sintasas/metabolismo , Pirazinas/química , Bazo/microbiología , Staphylococcus aureus/genética , Staphylococcus aureus/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA